• Aucun résultat trouvé

Degradable and biocompatible nanoparticles decorated with cyclic RGD peptide for efficient drug delivery to hepatoma cells in vitro.

N/A
N/A
Protected

Academic year: 2021

Partager "Degradable and biocompatible nanoparticles decorated with cyclic RGD peptide for efficient drug delivery to hepatoma cells in vitro."

Copied!
39
0
0

Texte intégral

(1)

HAL Id: hal-00861299

https://hal-univ-rennes1.archives-ouvertes.fr/hal-00861299

Submitted on 12 Sep 2013

HAL is a multi-disciplinary open access archive for the deposit and dissemination of sci-entific research documents, whether they are pub-lished or not. The documents may come from teaching and research institutions in France or abroad, or from public or private research centers.

L’archive ouverte pluridisciplinaire HAL, est destinée au dépôt et à la diffusion de documents scientifiques de niveau recherche, publiés ou non, émanant des établissements d’enseignement et de recherche français ou étrangers, des laboratoires publics ou privés.

Degradable and biocompatible nanoparticles decorated

with cyclic RGD peptide for efficient drug delivery to

hepatoma cells in vitro.

Pascal Loyer, Wahib Bedhouche, Zhi Wei Huang, Sandrine Cammas-Marion

To cite this version:

Pascal Loyer, Wahib Bedhouche, Zhi Wei Huang, Sandrine Cammas-Marion. Degradable and bio-compatible nanoparticles decorated with cyclic RGD peptide for efficient drug delivery to hep-atoma cells in vitro.. International Journal of Pharmaceutics, Elsevier, 2013, 454 (2), pp.727-37. �10.1016/j.ijpharm.2013.05.060�. �hal-00861299�

(2)

1

Degradable and Biocompatible Nanoparticles Decorated with Cyclic RGD Peptide for Efficient

1

Drug Delivery to Hepatoma Cells In Vitro

2

Pascal Loyer1, Wahib Bedhouche1, Zhi Wei Huang2,§, Sandrine Cammas-Marion2,*. 3

1

Inserm UMR S-991, Foie, Métabolismes et Cancer; Université de Rennes 1; Fédération de Recherche 4

de Rennes Biosit; CHU Rennes, 35033 Rennes, France. 5

2

UMR 6226 CNRS; Institut des Sciences Chimiques de Rennes; Université de Rennes 1; ENSCR, 6

Avenue du Général Leclerc, CS 50837, 35 708 Rennes Cedex 7, France. 7

§

Current address: UMR 6270 CNRS; Laboratoire Polymères Biopolymères Surfaces; Université de 8

Rouen, 76821 Mont Saint Aignan, France. 9

* Author to whom correspondence should be addressed

10

e-mail: sandrine.marion.1@ensc-rennes.fr; Tel: +33 2 23 23 81 09; Fax: +33 2 23 23 80 46. 11

12

Keywords: Biotinylated nanoparticles; site-specific targeting; anti-cancer drug encapsulation; 13

degradable poly(benzyl malate) derivatives; cyclic RGD peptide; HepaRG hepatoma cells. 14

15

ABSTRACT 16

Amphiphilic derivatives of poly(benzyl malate) were synthesized and characterized with the aim of 17

being used as degradable and biocompatible building blocks for the design of functional nanoparticles 18

(NPs). An anti-cancer model drug, doxorubicin, has been successfully encapsulated into the prepared 19

NPs and its release profile has been evaluated in water and in culture medium. NPs bearing biotin 20

molecules were prepared either for site-specific drug delivery via the targeting of biotin receptors 21

overexpressed on the surface of several cancer cells, or for grafting biotinylated cyclic RGD peptide 22

onto their surface using the strong and highly specific interactions between biotin and the streptavidin 23

protein. We have shown that this binding did not affect dramatically the physico-chemical properties 24

of the corresponding NPs. Cyclic RGD grafted fluorescent NPs were more efficiently uptaken by the 25

(3)

2 HepaRG hepatoma cells than biotinylated fluorescent NPs. Furthermore, the targeting of HepaRG 26

hepatoma cells with NPs bearing cyclic RGD was very efficient and much weaker for HeLa and HT29 27

cell lines confirming that cyclic RGD is a suitable targeting agent for liver cells. Our results also 28

provide a new mean for rapid screening of short hepatotropic peptides in order to design NPs showing 29

specific liver targeting properties. 30

31

1. Introduction

32

Nanotechnology, especially nanomedicine corresponding to the use of nanoparticles (NPs) in 33

biomedicine, is currently an ever growing scientific and technological domain [Psimidas et al., 2012; 34

Garanger et al., 2012]. The main reason for this unprecedented development relies on the aim to 35

improve both early detection and treatment of numerous pathologies such as cancers. The 36

encapsulation of a selected biologically active molecule into NPs might result in an increased drug 37

bioavaibility within solid tumors, arising from a decrease in its non-specific recognition by the 38

reticuloendothelial system (RES) and an improvement of its in vivo specific biodistribution, as well as 39

a minimized toxicity against healthy tissues and organs [Yan et al., 2012; Elsaesser et al., 2012; 40

Lamprecht, 2008]. Knowing that cancer, characterized by an abnormal and anarchical cell proliferation 41

within normal tissue, is a very complex disease and a major cause of mortality [Misra et al., 2010], the 42

development of efficient nanomedicine is thus a major challenge for public health [Reddy et al., 2011]. 43

In this context, several anti-cancer drug loaded NPs such as Doxyl® and Abraxane® have been 44

approved by the Food and Drug Administration (FDA) for clinical uses [Yan et al., 2012; Wang et al., 45

2012; Jain et al., 2010]. However besides these encouraging results, several challenges have to be 46

overcome in order to obtain NPs allowing highly efficient site-specific drug delivery. 47

The materials constituting the NPs have to respect very strict specifications: they must be (i) 48

biocompatible and non-toxic, (ii) (bio)degradable into non-toxic low molecular weight molecules or, at 49

least, bioassimilable after releasing the encapsulated drug, (iii) undetectable by the RES meaning 50

having stealth properties, (iv) adapted for carrying large amounts of drug that should be released in a 51

(4)

3 controlled manner at its site of action (targeting). Within this context, we have recently developed a 52

family of degradable non-toxic polymers derived from poly(malic acid), PMLA, which are able to 53

form well-defined NPs [Huang et al., 2012]. We have selected PMLA as macromolecular backbone 54

because this polymer, originally synthesized for application in the biomedical field, has been 55

successfully used as a platform in the synthesis of nanovectors [Huang et al., 2012; Cammas et al., 56

2000; Cammas-Marion et al., 2000; Osanai et al., 2000; Martinez Barbosa et al., 2004; Abdellaoui et 57

al., 1998] and macromolecular conjugates [Ding et al., 2010; Ljubimova et al., 2008; Fujita et al., 58

2007; Fujita et al., 2006]. PMLA is known to be non-toxic and degradable into malic acid under 59

physiological conditions [Vert et al., 1979] and its derivatives are accessible from naturally occurring 60

PMLA [Ljubimova et al., 2008] or by anionic ring-opening polymerization (ROP) of β-substituted β -61

lactones [Cammas et al., 1996; Cammas et al., 1993]. PMLA derivatives used for the formulation of 62

NPs were obtained by ROP of benzyl malolactonate (MLABe) in presence of either 63

tetraethylammonium benzoate, α-methoxy ω-carboxy poly(ethylene glycol) -PEG42-CO2H- or α-biotin

64

ω-carboxy poly(ethylene glycol) -Biot-PEG62-CO2H- as initiator [Huang et al., 2012]. Starting from

65

these three PMLA derivatives, we were able to obtain well-defined non-toxic NPs in which the 66

doxorubicin (Dox), and a fluorescent probe, the DiD oil, have been successfully encapsulated for in 67

vitro assays [Huang et al., 2012]. It is worth noting that PEG has been selected as hydrophilic block 68

because it is a well-known polymer conferring stealth properties at nanoparticles on which it is grafted 69

[Romberg et al., 2008]. On the other hand, biotin has been chosen firstly because it is a targeting agent 70

of certain cancer cells [Le Droumaguet et al., 2012; Patil et al., 2099; Kim et al., 2007] and secondly 71

because it is able to interact strongly with streptavidin [Yang et al., 2009] which is an important 72

property for our study as it will be explained afterwards in this paper. 73

Hepatocellular carcinoma (HCC) is the main primary malignant tumor of the liver representing 80 to 74

90% of liver tumors. It is the fifth most common tumor worldwide (5.4% of new cancer cases per year) 75

and the third in term of mortality (8.2% of all cancer death) [Parkin et al., 2005]. Early detection and 76

classification of HCC are crucial for the choice and effectiveness of therapeutic strategy. For small size 77

(5)

4 HCC, surgical treatment (resection and liver transplantation) is the most effective treatment [Hasegawa 78

et al., 2009; Mazzeferro et al., 2008; Ishikawa et al., 1992]. Palliative treatments such as 79

chemoembolization [Bernades-Genisson et al., 2003] and, more recently, chemotherapy using an 80

inhibitor of tyrosine kinase, Sorafenib (Nexavar®) are proposed to patients with advanced HCC. 81

Despites these advances, the therapeutic options for the treatment of HCC remains limited partly due 82

to the chemoresistance of liver tumors to conventional anti-tumor agents. Therefore, the use of 83

nanocarriers containing anti-tumor drugs has been envisaged for the treatment of HCC in order to 84

increase the intra-hepatic drug concentration while limiting the exposure of healthy tissues and side 85

effects [Reddy et al., 2011]. Several formulations are currently undergoing clinical trials in phase II 86

and III such as NPs of poly(alkyl cyanoacrylate) loaded with doxorubicin (Trandrug®) for HCC 87

treatment [Barraud et al., 2005]. In a first step, the passive accumulation of nanocarriers in the RES 88

cells (endothelial and Kupffer cells) [Lanaerts et al., 1984] was utilized for liver targeting with a real 89

relevance for diseases involving liver Kupffer cells such as parasitic diseases [Alving et al., 1978]. 90

Conversely, liver targeting based on the nanocarrier’s uptake by Kupffer cells has the major drawback 91

of allowing only a low hepatic accumulation of nanocarriers since Kupffer cells represent only a few 92

percent of liver cell volume against 90% for hepatocytes. In addition, the accumulation of nanocarriers 93

in Kupffer cells does not target the cells responsible for HCC thus limiting the use of this approach in 94

this case. Therefore, to overcome this drawback, active hepatocyte targeting has been studied. Most of 95

the proposed strategies are based on the binding of NPs to the asialoglycoprotein receptors [Wu et al., 96

2002]. These NPs are usually liposomes incorporating glycosylated proteins or galactose/lactose linked 97

to lipophilic anchors [Wu et al., 2002]. To date, very few nanovectors, especially polymer-based NPs, 98

carrying peptides with high tropism for the liver have been developed [Reddy et al., 2011]. 99

In this paper, we report, first, the monitoring of Dox release from NPs prepared from PEG42

-b-100

PMLABe or Biot-PEG62-b-PMLABe in water and in culture medium at 37°C. Second, we have grafted

101

fluorescein amine (FA) molecule at the free end of the PMLABe block of PEG42-b-PMLABe or

Biot-102

PEG62-b-PMLABe block copolymers in order to obtain fluorescent NPs for in vitro cell uptake assays.

(6)

5 Starting from the corresponding fluorescent NPs, we have studied the influence of the nature of 104

molecules localized at NPs’ surfaces [PEG42, Biot-PEG62 or Arginine-Glycine-Aspartic acid (RGD)

105

peptide-Biot-Streptavidin-Biot-PEG62] on their internalization into cells in vitro and demonstrated that

106

the uptake by HepaRG hepatoma cells is considerably enhanced by the grafting of the RGD peptide 107

onto NPs. These results also show that these biotinylated NPs can be useful tools for rapid in vitro 108

screen and selection of highly hepatotropic peptides with the ultimate goal to design nanomedicine 109

targeting hepatocytes from HCC in vivo. 110

111

2. Materials and methods

112

2.1. Materials 113

All chemicals were used as received. Anhydrous THF was obtained by distillation over 114

sodium/benzophenone under N2 atmosphere.

115

Three cell lines have been selected within the frame of this project: the HepaRG hepatoma cells 116

[Gripon at al., 2002; Laurent et al., 2010;], the colorectal adenocarcinoma cell line HT29 [Fogh et al., 117

1975] and the cervical cancer cell line HeLa [Rahbari et al., 2009]. 118

119

2.2. Apparatus 120

Nuclear magnetic resonance spectra (1H NMR) were recorded on a Brucker ARX 400 instrument (1H 121

at 400 MHz). Data are reported as follows: chemical shift (multiplicity, number of hydrogen). The 122

chemical shifts (δ) are reported as parts per million (ppm) referenced to the appropriate residual solvent 123

peak. Abbreviations are as follows: s (singlet), d (doublet), t (triplet), q (quartet), dd (doublet of 124

doublet), m (multiplet). 125

The size (average diameter obtained by the cumulant result method), polydispersity and zeta potential 126

of the formulations were measured by dynamic light scattering using a Delsa™ Nano Beckman 127

Coulter apparatus at 25°C. 128

UV spectra were recorded on a Secoman apparatus at 485 nm. 129

(7)

6 2.3. Synthesis of PEG42-b-PMLABe and Biot-PEG62-b-PMLABe

130

The monomer, the benzyl malolactonate (MLABe), was synthesized from DL-aspartic acid according 131

to the previously reported synthesis [Cammas et al., 1996]. The PEG42-b-PMLABe and Biot-PEG62

-b-132

PMLABe block copolymers were obtained by anionic ring opening polymerization of MLABe in 133

presence of, respectively, α-methoxy ω-carboxy poly(ethylene glycol), PEG42-CO2H, and α-biotin ω

-134

carboxy poly(ethylene glycol), Biot-PEG62-CO2H, as initiators following a protocol described

135

elsewhere [Huang et al., 2012]. 136

137

2.4. Grafting of fluorescein amine (FA) 138

2.4.1 Grafting of FA on the PEG42-b-PMLABe block copolymer (Scheme 1)

139

The PEG42-b-PMLABe block copolymer (500 mg) was dissolved into 1 mL of anhydrous CH2Cl2

140

under nitrogen atmosphere. To this solution were added 5 mg (1eq.) of N,N’-diclohexylcarbodiimide 141

(DCC) dissolved in 1 mL of anhydrous CH2Cl2 followed by 7.5 mg (1eq.) of N-hydroxysuccinimide

142

(NSH) dissolved in 1 mL of anhydrous CH2Cl2. The mixture was stirred under nitrogen atmosphere for

143

24 hours at room temperature (RT). The resulting PEG42-b-PMLABe-NHS was precipitated in cold

144

heptane. After the elimination of the supernatant, the precipitate was dissolved in CH2Cl2 and the

145

solution was filtrated on celite. The CH2Cl2 was eliminated under vacuum and the activated block

146

copolymer polymer (480 mg) was obtained with 96% yield. The activated block copolymer (480 mg) 147

was then dissolved in 1 mL of anhydrous CH2Cl2. To this solution was added 12 mg of FA (1eq.)

148

solubilised into a mixture of 1 mL of anhydrous CH2Cl2 and1 mL of acetone HPLC grade under

149

nitrogen atmosphere. After stirring at RT for 24 hours, the solution containing the PEG42

-b-PMLABe-150

FA block copolymer was precipitated into cold heptane. After removing the supernatant, the 151

precipitate was dissolved into DMSO. This DMSO solution was poured into a dialysis bag (MWCO 152

3,500 Da) and the dialysis was conducted during 8 hours against DMSO. The solution contained into 153

the dialysis bag was lyophilized and the PEG42-b-PMLABe-FA block copolymer was recovered with

154

60% yield. The polymer was characterized by 1H NMR in deuterated DMSO. 155

(8)

7 1 H NMR (d6-DMSO, δ ppm): 2.92 (s, 2nH, CO2CH2C6H5), 3.32 (m, 4mH (m=42), (CH2CH2O)42), 5.10 156 (m, 2nH, CHCH2CO2), 5.42 (m, 1nH, CHCH2CO2), 6.50-7.00 (m, 9H, FA); 7.31 (m, 5nH, 157 CO2CH2C6H5). 158

MNMR = 11,000 g/mol for the PMLABe block 159

160

2.4.2 Grafting of FA on the Biot-PEG62-b-PMLABe block copolymer (Scheme 1)

161

The grafting of FA on the Biot-PEG62-b-PMLABe block copolymer was realized as described above.

162

The Biot-PEG62-b-PMLABe-FA block copolymer was obtained with 58% yield and characterized by

163 1 H NMR in DMSO. 164 1H NMR (d6-DMSO, δ ppm): 2.92 (s, 2nH, CO 2CH2C6H5), 3.37 (m, 4mH (m=62), (CH2CH2O)62), 5.07 165 (m, 2nH, CHCH2CO2), 5.41 (m, 1nH, CHCH2CO2), 6.50-7.00 (m, 9H, FA); 7.26 (m, 5nH, 166

CO2CH2C6H5). Peaks corresponding to the biotin are either under peaks correspoonding to the PEG 167

and PMLABe blocks or too small to be detectable on the NMR spectrum. 168

MNMR = 6,000 g/mol for the PMLABe block. 169

170

2.5. Preparation of NPs 171

2.5.1. Dox encapsulation and release from NPs 172

The protocol for Dox encapsulation and for the monitoring of its release was the same whatever the 173

nature of the block copolymer constituted the NPs. The encapsulation of Dox into the NPs was realised 174

as described previously [Huang et al., 2012]. Briefly, the Dox hydrochloride (Dox,HCl, Sigma) was 175

encapsulated into the two kinds of NPs during the nanoprecipitation procedure. The selected polymer 176

(5 mg) was dissolved in acetone (1 mL). Two hundred µ L of a Dox solution [1.5 mg of Dox,HCl 177

solubilised in 0.6 mL of a mixture of chloroform (6 mL) and NEt3 (23 µ L)] were added to the polymer

178

solution. This mixture was then nanoprecipitated into 2 mL of water under vigorous stirring. After 179

organic solvent evaporation, the unloaded Dox was removed by ultracentrifugation at 15,000 g at 15°C 180

for 7 min using filter with an exclusion limit 10,000 Da. The filters were returned and centrifuged for 1 181

(9)

8 min at 1,000 g at 15°C. The volume of the recovered solutions was completed to 2 mL with distilled 182

water in order to obtain a final concentration in NPs of 2.5 g/L. The concentration of loaded Dox was 183

evaluated by UV at 485 nm, as described elsewhere [Huang et al., 2012; Cammas et al., 1995]. Briefly, 184

200 µ L of Dox-loaded NPs were dissolved into 800 µ L of DMF and the resulting solutions were 185

analyzed by UV at 485 nm. The absorbance of Dox encapsulated into NPs was converted into a 186

concentration using a calibration curve and the encapsulation efficiency (e.e.) was calculated using the 187

following equation: 188

189 190

The characteristics of the Dox-loaded NPs (diameter, polydispersity index and zeta potential) were 191

measured using the Delsa™ Nano Beckman Coulter apparatus (Table 1). 192

The Dox release from both kinds of NPs were monitoring by dialysis in water and in culture medium 193

at 37°C. The protocol used in both cases was identical. Two mL of the Dox-loaded NPs solution were 194

placed into a dialysis bag (MWCO = 3,500 Da); this bag was then incubated into 40 mL of water or 195

culture medium maintained at 37°C. After different incubation time, from 30 min up to 72 hours, 2 mL 196

of the outside solution are taken and replaced by 2 mL of fresh water or culture medium. For each 197

sample, 200 µ L of the collected outside solution were analyzed by UV at 485 nm and the quantity of 198

Dox was determined thanks to calibration curves previously realized in water and in culture medium 199 by UV measurements at 485 nm. 200 201 2.5.2. Preparation of fluorescent NPs 202

The fluorescent NPs were prepared by the nanoprecipitation technique as previously described 203

[Thioune et al., 1997; Huang et al., 2012]. Briefly, the mixture of PEG42-b-PMLABe-FA (2.5 mg) and

204

PEG42-b-PMLABe (2.5 mg) or Biot-PEG62-b-PMLABe-FA (2.5 mg) and Biot-PEG62-b-PMLABe (2.5

205

mg) were dissolved in 1 mL of acetone. This solution is added to 2 mL of water under vigorous 206

stirring. The organic solvent (acetone) was then evaporated under vacuum and the final volume was 207

(10)

9 completed to 2 mL with fresh water. The final concentration in block copolymers under NPs’ form was 208

2.5 g/L. The solutions containing the fluorescent NPs were characterized by dynamic light scattering 209

using a Delsa™ Nano Beckman Coulter apparatus at 25°C (Table 2). 210

211

2.5.3. Grafting of the cyclic RGD peptide 212

The selected biotinylated peptide, the cyclic Biot-RGD peptide (Eurogentec, Belgium), was grafted 213

onto the biotinylated NP’s surfaces via the streptavidin (Strept, AnaSpect, Eurogentec, Belgium) 214

protein. An aqueous solution of Biot-RGD peptide was prepared at a final concentration of 2.9 mM. In 215

parallel, an aqueous solution of streptavidin was also prepared with a final concentration of 178 µM. 216

The Biot-RGD peptide (2.7 µ L) and the streptavidin (22 µ L) solutions were mixed in a final volume of 217

50 µ L (H2O qsp) and incubated for 1 hour at 4°C. Then, the Biot-PEG62-b-PMLABe-FA or

Biot-218

PEG62-b-PMLABe based NP’s solution (35 µ L), previously prepared, was added to a final volume of

219

100 µ L (H2O qsp). This mixture was incubated for 1 hour and diluted to a final volume of 1 mL in

220

culture medium for final concentrations of the Biot-RGD peptide at 8µM, the streptavidin at 4µM and 221

polymers at 4µM. In order to demonstrate that both the Biot-RGD peptide grafting and the dilution 222

have no influence on NP’s characteristics, we analyzed the NPs formed in the conditions described 223

above by DLS (Table 3). 224

225

2.6. Cell uptake assays 226

The cell lines, HT29 [Fogh et al., 1975] and HeLa [Rahbari et al., 2009], were cultured as described in 227

the literature. The HepaRG cell line was cultured in the medium William’s E (Lonza) supplemented 228

with 2mM of glutamine (Gibco), 5 mg/L of insulin (Sigma), 10-5 M hydrocortisone hemisuccinate and 229

10% of fetal calf serum (Lonza) [Gripon at al., 2002; Laurent et al., 2010]. During the sub-culturing, 230

2.106 cells were seeded in a 75 cm3 flask. The medium was renewed every 48 hours. The sub-culturing 231

was realized by trypsinization every 2 weeks in order to maintain the progenitor phenotype. For an 232

(11)

10 optimal differentiation, the cells were maintained at confluence after the two weeks and the medium 233

was supplemented with 2% of dimethylsulfoxide (DMSO) [Laurent et al., 2013]. 234

For the cell uptake assays, the 24 wells culture plates were seeded with the selected cell line (HepaRG, 235

HT29 or HeLa) with 105 cells per well. Then the NPs’ preparations (4µM of block copolymer under 236

NPs’ form ± 4 µM streptavidin and 8 µM Biot-RGD) or a negative control (buffer without NPs or non 237

fluorescent NPs) were added to the wells. For the competitive experiments, the cells were pre-treated 238

with an excess of free RGD peptide (32 µM). 239

The cells were incubated from 1 to 24 hours. After incubation, the culture medium was removed; the 240

cell monolayers were washed with PBS before the observation by fluorescence microscopy (Zeiss 241

inverted microscope, analysis software AxioVision). Then the cells were detached with trypsin and 242

analyzed by flow cytometry (FACSCalibur Becton Dikinson) to quantify the fluorescence (Channel 243

FL1H) emitted by the fluorescent NPs captured by the cells. Cytometry data were analyzed using 244

CellQuest software (Becton Dikinson). 245

246

3. Results and Discussion

247

In order to further characterize the PMLABe based NPs as drug nanocarriers for applications in 248

nanomedicine, we (i) studied the release of Dox in water and culture medium at 37°C, (ii) grafted a 249

fluorescent probe at the free end of the hydrophobic PMLABe block for studying in vitro cellular 250

uptake and (iii) evaluated the possibility to build a molecular scaffold by grafting the cyclic RGD-251

biotinylated peptide onto biotinylated NPs via the streptavidin as a bridging factor and determine the 252

impact of the RGD peptide addition on cell uptake. 253 254 Figure 1 255 256 257 258

(12)

11 3.1. Dox encapsulation and release from NPs

259

Both Dox-loaded PEG42-b-PMLABe and Biot-PEG62-b-PMLABe based NPs have been prepared by

260

the nanoprecipitation technique and have been characterized by DLS. Table 1 collects the results 261

obtained for PEG42-b-PMLABe and Biot-PEG62-b-PMLABe based NPs. Initial Dox content in both

262

NPs determined by UV at 485 nm as described previously [Huang et al., 2012] showed an 263

encapsulation efficiency ranging from 32 to 36%. 264

265

Table 1 266

267

The release of Dox from both PEG42-b-PMLABe and Biot-PEG62-b-PMLABe NPs in water and in

268

culture medium was realized at 37°C by dialysis. Aliquots of the external solution (outside the dialysis 269

bag) were collected after various incubation times and analyzed by UV at 485 nm and confirmed that 270

the Dox was encapsulated with an efficiency of nearly 35% (Table 1). As shown in figure 2, Dox 271

release profiles from both types of NPs in water (Figure 2, A) and in the culture medium (Figure 2, B) 272

are quite similar. 273

274

Figure 2 275

276

The release, expressed as a percentage of the total amount of encapsulated Dox, is nearly 5% after one 277

hour of incubation. This release accelerates after the second hour to reach 40% after 6 hours of 278

incubation in both water and culture medium. Then the release reaches a plateau around 55 to 60% 279

from 24 to 72 hours of incubation. The fast release of Dox within the first hours is probably due to the 280

Dox absorbed in the hydrophilic PEG corona. Indeed, Dox is known to be an amphiphilic molecule 281

which is therefore spread from the hydrophilic corona to the surface of the hydrophobic core of the 282

nanoparticles. However to conclude regarding the exact location of the Dox in the PEG-b-PMLABe 283

forming NPs, it will be necessary to realize further experiments such as X-ray measurements. 284

(13)

12 Nevertheless, these results are quite encouraging because we are able to encapsulate substantial 285

amount of this drug and the release profile is in agreement with the Dox amphiphilic nature. 286

287

3.2. Grafting of fluorescein amine and cellular uptake assays 288

Besides the use of biotin as a well-known targeting agent of cancer cells [Yang et al., 2009], we aimed 289

at developing a versatile procedure to screen for more specific targeting agents, especially short 290

peptides, towards transformed hepatocytes from hepatocellular carcinoma (HCC). 291

Our goal is to select peptides exhibiting a remarkably high tropism for the hepatocytes as targeting 292

agents and to graft them at the surface of PMLA derivatives-based NPs in order to achieve an 293

optimized uptake of NPs by the hepatocytes. In a first step, we wish to screen, rapidly and in a simple 294

manner without engaging more organic chemistry, a large number of peptides which potentially show 295

a strong hepatotropism to select the most efficient ones. For that purpose, we used the non-covalent 296

binding of selected peptides via the strong biotin-streptavidin affinity as shown by Figure 3 [Yang et 297 al., 2009]. 298 299 Figure 3 300 301

In a first step to determine whether such a molecular scaffold could allow the production of NPs and 302

could be used for cell uptake in vitro, we have selected biotinylated cyclic RDG (Arg-Gly-Asp- DTyr-303

Lys-Biotin) peptide, known for interacting with integrin proteins well expressed in the liver and even 304

more in tumors rich in extracellular matrix [Jiang et al., 2011]. This biotinylated RGD peptide has been 305

introduced after the formation of biotinylated NPs through streptavidin interactions [Yang et al., 2009]. 306

Moreover, in order to follow the in vitro cellular uptake of the NPs, we have synthesized fluorescein 307

amine grafted PEG42-b-PMLABe and Biot-PEG62-b-PMLABe block copolymers. As shown by scheme

308

1, the fluorescein amine (FA) was successfully grafted at the free carboxylic acid end of the PMLABe 309

(14)

13 block activated with N-hydroxysuccinimide (NHS) without modifying the structure of block 310 copolymers. 311 312 Scheme 1 313 314

After purification by dialysis allowing the elimination of unreacted FA and low molecular weight side 315

products, both block copolymers were characterized by 1H NMR in deuterated DMSO. The 1H NMR 316

spectra allowed us to conclude that the structures of FA-modified block copolymers were in agreement 317

with the expected ones and that the molecular weights of PMLABe blocks calculated from the 1H 318

NMR spectra were identical to the ones of initial materials. 319

Starting from a mixture of the fluorescent block copolymers and the non-fluorescent ones (50/50 wt%), 320

we have then prepared the corresponding fluorescent NPs using the nanoprecipitation method [Huang 321

et al., 2012; Thioune et al., 1997]. The obtained NPs were characterized by dynamic light scattering. 322

As shown by results gathered in table 2, the presence of FA molecules at the end of the PMLABe 323

block has no significant influence on the NP’s diameters and polydispersity indices. 324

325

Table 2 326

327

Biotinylated cyclic RGD peptide was then associated with NPs formed by a mixture of Biot-PEG62

-b-328

PMLABe-FA and Biot-PEG62-b-PMLABe (50/50 wt%) using streptavidin, a tetrameric protein of 56

329

KDa purified from the bacterium Streptomyces avidinii, as an intermediate link between the biotin 330

localized on the surface of preformed polymeric NPs and the biotinylated peptide (Figure 3). The 331

dissociation constant (Kd) of the biotin/streptavidin complex is on the order of 10-15 mol/L, ranking 332

among the strongest known non-covalent interactions [Yang et al., 2009]. The non-covalent binding of 333

RGD modified fluorescent NPs was realized by first mixing the biotinylated RGD peptide with the 334

streptavidin followed by the addition of this complex to biotinylated fluorescent NPs. The relative 335

(15)

14 amounts of block copolymers constituting the NPs, RGD peptide and streptavidin can have a 336

significant influence on the cell capture. Therefore, different amounts of block copolymer constituting 337

the NPs, RGD peptide and streptavidin were tested and the cell uptake was measured by fluorescent 338

microscopy and flow cytometry (Data not shown); the best results were obtained with the following 339

conditions: 4 µM of block copolymers under NP’s form, 4 µM of streptavidin and 8 µM of 340

biotinylated RGD peptide. 341

The diameter and polydispersity index of unmodified and modified NPs were measured by DLS in 342

order to demonstrate that the addition of RGD-Biot-streptavidin construct and the dilution had no 343

influence on the properties of the corresponding NPs. As shown by results gathered in table 3, the 344

dilution of Biot-PEG62-b-PMLABe NPs either in PBS or in culture medium with or without serum

345

does not have a significant influence on both the diameter and the polydispersity index values, 346

meaning that the polymers constituting the NPs are still associated under NP’s form. 347

348

Table 3 349

350

The addition of the Strep-Biot-RGD construct on the biotinylated NPs has led to a moderated increase 351

in the NPs’ diameter and polydispersity indices. For NPs resuspended either in PBS or culture medium 352

with serum the emergence of a second peak centered at 600 nm and 970 nm, respectively, was 353

observed. Such results indicate that addition of Strep-Biot-RGD construct does not lead to the 354

destabilization of the NPs but rather a moderate formation of aggregates. However, such limited 355

modifications were considered acceptable for cell uptake in vitro assays. It is important to note that 356

RGD modified NPs were stable for at least 48 hours when stored at room temperature as shown by 357

values of diameters and polydispersity indices unchanged (measures realized by DLS, data not shown). 358

We then realized in vitro assays of cell captation using the human hepatoma HepaRG cell line [Laurent 359

et al., 2010; Gripon et al., 2002]. HepaRG cells were incubated for 24 hours in the presence of 360

biotinylated NPs modified or not with the RGD-Biot-Streptavidin construct with final concentrations 361

(16)

15 in culture media of 4 µM of block copolymers, 4 µM of streptavidin and 8 µM of RGD peptide. The 362

cell uptake was studied by Facs and fluorescent microscopy (Figure 4). 363

364

Figure 4 365

366

The Facs analysis showed that the peak of fluorescence in cells incubated with the Biot-PEG62

-b-367

PMLABe-FA NPs had significantly shifted on the right compared to fluorescence in control cells 368

incubated with non fluorescent NPs demonstrating that most of the cells contained fluorescent NPs. 369

However, the mean of fluorescence remained low (~30 arbitrary units) for Biot-PEG62-b-PMLABe

370

NPs and Biot-PEG62-b-PMLABe-FA NPs, respectively, compared to the mean for cells incubated with

371

non-fluorescent NPs. The fluorescent microscopy confirmed that HepaRG cell uptake of Biot-PEG62

-372

b-PMLABe-FA NPs was very limited compared to negative control cells exposed to the non-373

fluorescent NPs demonstrating that the biotin did not trigger a strong captation. Addition of the RGD 374

peptide led to strong increase in the fluorescence level within HepaRG cells reaching a mean of ~1500 375

U.A and with over 90% of positive cells (Figure 4). These data demonstrate that the addition of the 376

RGD peptide onto NPs has considerably enhanced the cell uptake of the NPs. 377

We then followed the time course of uptake by HepaRG cells of NPs formed by Biot-PEG62

-b-378

PMLABe-FA or Biot-RGD-Strept-Biot-PEG62-b-PMLABe-FA by flow cytometry analysis at 2, 6, 14

379

and 24 hours of incubation. As shown by figure 5, the results of mean of fluorescence intensity 380

indicated a very rapid uptake within the first 6 hours which continued slower until 24 hours for RGD-381

Biot-Strept-Biot-PEG62-b-PMLABe-FA based NPs (Figure 5, red curve), while no change in the

382

fluorescence intensity has been observed for the uptake of Biot-PEG62-b-PMLABe-FA based NPs

383

(Figure 5, blue curve). 384

385

Figure 5 386

(17)

16 Furthermore, we have evaluated the specificity of the uptake of RGD modified fluorescent NPs by 388

HepaRG cells by comparing the efficiency of the uptake in absence or presence of an excess of free 389

RGD peptide at a concentration of 34 µM (Figure 6). The results of fluorescence intensity measured by 390

flow cytometry indicated that the pre-incubation of HepaRG cells with free RGD peptide before the 391

addition of RGD modified fluorescent NPs strongly inhibited the cell uptake. Therefore, we can 392

conclude that the presence of the RGD peptide at the NP’s surfaces is responsible for the increase in 393

HepaRG cell uptake of NPs. 394

395

Figure 6 396

397

In final experiment, we compared the efficiency of the uptake of RGD modified fluorescent NPs by 398

three different cell lines: the hepatoma HepaRG, the colon HT29 and the cervical HeLa cancer cells 399 (Figure 7). 400 401 Figure 7 402 403

The three cell lines showed low levels of fluorescence following incubation with Biot-PEG62

-b-404

PMLABe-FA NPs for 24 hours demonstrating that these biotinylated NPs are poorly uptaken by cells 405

from different tissue origin. Importantly, the three cell types exhibited very different uptake of the 406

RGD-Biot-Strept-Biot-PEG62- b-PMLABe-FA formed NPs. The number of positive HT29 cells is very

407

low (< 5%) while about 90% of HeLa cells captured the RGD modified fluorescent NPs (Figure 7). 408

However, the degree of uptake (fluorescence intensity) in HeLa cells is much lower than the one 409

observed for HepaRG cells (Figure 7). This result suggested that the number of membrane receptors 410

binding RGD peptide (integrin family members) on HeLa cells is sufficient to trigger the uptake of 411

NPs by most of the cells. However, the density of these receptors onto HeLa cells might be lower than 412

(18)

17 onto HepaRG cells leading to a higher degree of NPs captation by the hepatoma cells than the HeLa 413 cells. 414 415 4. Conclusions 416

In this study, we have demonstrated a rapid release of the Dox from PEG42-b-PMLABe and

Biot-417

PEG62-b-PMLABe NPs over the first 6 hours. However, the NPs, which are stable in water and culture

418

medium for several days, entrapped a fraction of the drug that is slowly released over several days. 419

Then, we characterized the cell uptake of PEG42-b-PMLABe and Biot-PEG62-b-PMLABe NPs using

420

fluorescein amine modified polymers. We demonstrated that biotinylated NPs modified with the cyclic 421

RGD peptide significantly increase the uptake by HepaRG cells in comparison to NPs formed by Biot-422

PEG62-b-PMLABe-FA without peptide. We have also proved that this capture was dependent on the

423

presence of the RGD peptide because the addition of an excess of free RGD peptide strongly inhibited 424

the uptake. In addition, our results indicate that the RGD peptide presents a real tropism for liver cells 425

since its uptake by HeLa and HT29 cells was much lower than the uptake by HepaRG cells. These 426

results should be complemented by a larger study including other liver and non-hepatic cell lines as 427

well as primary cells such as endothelial cells and normal hepatocytes. Our study also provides a proof 428

of concept for the use of the versatile molecular scaffold presented in Figure 3 in order to screen for 429

other short peptides targeting hepatocytes and HCC. Since we have shown that it was possible to use 430

the streptavidin to graft a biotinylated peptide onto the biotin present at the surface of Biot-PEG62

-b-431

PMLABe based NPs, we are evaluating a number of peptides without having to engage additional 432

chemistry. The most efficient peptides will be then grafted at the end of the hydrophilic PEG block to 433

avoid the use of immunogenic streptavidin for further in vivo biodistribution assays. 434

435

Acknowledgments

436

We would like to thank Denise Glaise for culturing the HepaRG cells. This work was supported by 437

Inserm, CNRS, the University of Rennes 1 (Défis émergents-2012) and les comités départementaux de 438

(19)

18 la Ligue contre le Cancer du GrandOuest: comités 29, 35 et 53. Z.H. H. thanks the Région Bretagne 439

and the European University of Bretagne (UEB) for a Ph.D. grant and a 4 months mobility fellowship, 440 respectively. 441 442 References 443

Abdellaoui, K., Boustta, M., Vert, M., Morjani, H., Manfait, M. 1998. Metabolite-derived artificial 444

polymers designed for drug targeting, cell penetration and bioresorption., Eur. J. Pharm. Sci. 6, 61-73. 445

446

Alving, C.R., Steck, E.A., Chapman, W.L. Jr., Waits, V.B., Hendricks, L.D., Swartz, G.M. Jr., Hanson, 447

W.L. 1978. Therapy of leishmaniasis: Superior efficacies of liposome-encapsulated drugs., Proc. Natl. 448

Acad. Sci. U S A 75(6), 2959-2963. 449

450

Barraud, L., Merle, P., Soma, E., Lefrançois, L., Guerret, S., Chevallier, M., Dubernet, C., Couvreur, 451

P., Trépo, C., Vitvitski, L. 2005. Increase of doxorubicin sensitivity by doxorubicin-loaded into 452

nanoparticles for hepatocellular carcinoma cells in vitro and in vivo., J. Hepatol. 42(5), 736-743. 453

454

Bernades-Genisson, V., Bernadou, J., Berque-Bestel, I., Brion, J.D., Couquelet, J., Cussac, M., Debert, 455

M., Duval, O., Giorgi-Renault, S., Huet, J., Lacroix, R., Laronze, J.Y., Le Baut, G., Loiseau, P., 456

Nuhrich, A., Plat, M., Poisson, J., Robert-Piessard, S., Tournaire-Arllano, C., Uriac, P. 2003. In « 457

Traité de chimie thérapeutique », vol. 6, « Médicaments anti-tumoraux et perspectives dans les 458

traitements des cancers ». Edition TEC et DOC. 3-23. 459

460

Cammas, S., Renard, I., Boutault, K., Guérin, Ph. 1993. A novel synthesis of optically active 4-461

benzyloxy- and 4-alkyloxycarbonyl-2 oxetanones., Tetrahedron Asymmetry 4(8), 1925-1930. 462

(20)

19 Cammas, S., Nagasaki, Y., Kataoka, K. 1995. Heterobifunctional Poly(ethylene oxide) : Synthesis of 464

α-methoxy-ω-amino and α-hydroxy-ω-amino PEOs with the same molecular weights., Bioconjugate 465

Chem. 6, 226-230. 466

467

Cammas, S., Renard, I., Langlois, V., Guérin, Ph. 1996. Poly(β-malic acid): obtaining of high 468

molecular weights by improvement of the synthesis route., Polymer 37(18), 4215-4220. 469

470

Cammas, S., Béar, M.M., Harada, A., Guérin, Ph., Kataoka, K. 2000. New macromolecular micelles 471

based on degradable amphiphilic block copolymers., Macromol. Chem. Phys. 201(3), 355-364. 472

473

Cammas-Marion, S., Guérin, Ph. 2000. 4-Alkyloxycarbonyl-2-oxetanones and 3-alkyloxycarbonyl-2-474

oxetanones as versatile chiral precursors in the design of functionalized polyesters with controlled 475

architecture., Designed Monomers and Polymers (DMP) 3(1), 77-93. 476

477

Ding, H., Inoue, S., Ljubimov, L.V., Patil, R., Portilla-Arias, J., Hu, J., Konda, B., Wawrowsky, K.A., 478

Fujita, M., Karabalin, N., Sasaki, T., Black, K.B., Holler, E., Ljubimova, J.Y. 2010. Inhibition of brain 479

tumor growth by intravenous poly(β-L-malic acid) nanobioconjugate with pH-dependent drug release., 480

Proc. Natl. Acad. Sci. USA 107, 18143-18148. 481

482

Elsaesser, A., Howard, C.V. 2012. Toxicology of nanoparticles. Advanced Drug Delivery Reviews 64, 483

129-137. 484

485

Fujita, M., Khazenzon, N.M., Ljubimov, A.V., Lee, B.S., Virtanen, I., Holler, E., Black, K.L., 486

Ljubimova, J.Y. 2006. Inhibition of laminin-8 in vivo using a novel poly(malic acid)-based carrier 487

reduces glioma angiogenesis., Angiogenesis 9, 183-191. 488

(21)

20 Fujita, M., Lee, B.S., Khazenzon, N.M., Penichet, M.L., Wawrowsky, K.A., Patil, R., Ding, H., 490

Holler, E., Black, K.L., Ljubimova, J.Y. 2007. Brain tumor tandem targeting using a combination of 491

monoclonal antobodies attached to biopoly(β-L-malic acid)., J. Controlled Release 122 (3), 356-363. 492

493

Fogh, J., Trempe, G., 1975. In “Human Tumor Cells in Vitro”, Ed. J. Fogh, Plenum, New York 115-494

140. 495

496

Garanger, E., Lecommandoux, S., 2012. Towards Bioactive Nanovehicles Based on Protein Polymers., 497

Angew Chem. Int. Ed. 51, 3060-3062. 498

499

Gripon, P., Rumin, S., Urban, S., Le Seyec, J., Glaise, D., Cannie, I., Guyomard, C., Lucas, J., Trepo, 500

C., Guguen-Guillouzo, C. 2002. Infection of a human hepatoma cell line by hepatitis B virus., Proc. 501

Natl. Acad. Sci. USA 99(24), 15655-15660. 502

503

Hasegawa, K., Kokudo, N. 2009. Surgical treatment of hepatocellular carcinoma., Surg. Today 39(10), 504

833-843. 505

506

Huang, Z.W., Laurent, V., Chetouani, G., Ljubimova, J.Y., Holler, E., Benvegnu, T., Loyer, P., 507

Cammas-Marion, S. 2012. New functional degradable and bio-compatible nanoparticles based on 508

poly(malic acid) derivatives for site-specific anti-cancer drug delivery., Int. J. Pharm. 423, 84-92. 509

510

Ishikawa, T. 1992. The ATP-dependent glutathione S-conjugate export pump., Trends Biochem. Sci. 511

17(11), 463-468. 512

513

Jain, R.K., Stylianopoulos, T. 2010. Delivering nanomedicine to solid tumors., Nat. Rev. Clin. Oncol. 514

7(11), 653-664. 515

(22)

21 Jiang, X., Sha, X., Xin, H., Chen, L., Gao, X., Wang, X., Law, K., Gu, J., Chen, Y., Jiang, Y., Ren, X., 516

Ren, Q., Fang, X. 2011. Self-aggregated pegylated poly(trimethylene carbonate) nanoparticles 517

decorated with c(RGDyK) peptide for targeted paclitaxel delivery to integrin-rich tumors., 518

Biomaterials 32, 9457-9469. 519

520

Kim, S.Y., Cho, S.H., Lee, Y.M., Chu, L.Y. 2007. Biotin-conjugated block copolymeric nanoparticles 521

as tumor-targeted drug delivery systems. Macromol. Res. 15, 646-655. 522

523

Lamprecht, A. 2008. In : Nanotherapeutics – Drug Delivery Concept in Nanoscience, Publisher: Pan 524

Standford Publishing Pte. Ltd., World Scientific Publishing Co. Pte. Ltd., Singapore. 525

526

Lanaerts, V., Nagelkerke, J.F., Van Berkel, T.J., Couvreur, P., Grislain, L., Roland, M., Speiser, P. 527

1984. In vivo uptake of polyisobutyl cyanoacrylate nanoparticles by rat liver Kupffer, endothelial, and 528

parenchymal cells., J. Pharm. Sci. 73(7), 980-982. 529

530

Laurent, V., Fraix, A., Montier, T., Cammas-Marion, S., Ribault, C., Benvengu, T., Jaffres, P-A. 531

Loyer, P. 2010. Highly efficient gene transfer into hepatocyte-like cells: new means for drug 532

metabolism and toxicity studies., Biotechnol. J. 5, 314-320. 533

534

Laurent, V., Glaise, D., Nübel, T., Gilot, D., Corlu, A., Loyer, P. 2013. Highly Efficient SiRNA and 535

Gene Transfer into Hepatocyte-Like HepaRG Cells and Primary Human Hepatocytes. Methods Mol 536

Biol. 987, 295-314. 537

538

Le Droumaguet, B., Nicolas, J., Brambilla, D., Mura, S., Maksimenko, A., De Kimpe, L., Salvati, E., 539

Zona, C., Airoldi, C., Canovi, M., Gobbi, M., Noiray, M., La Ferla, B., Nicotra, F., Scheper, W., 540

Flores, O., Masserini, M., Andrieux, K., Couvreur, P. 2012. Versatile and efficient targeting using a 541

(23)

22 single nanoparticulate platform: application to cancer and Alzheimer’s disease. ACS Nano 6, 5866-542

5879. 543

544

Ljubimova, J.Y., Fujita, M., Khazenzon, N.M., Lee, B.S., Wachsmann-Hogiu, S., Farkas, D.L., Black, 545

K.L., Holler, E. 2008. Nanoconjugate based on polymalic acid for tumor targeting., Chem. Biol. 546

Interact. 171, 195-203. 547

548

Martinez Barbosa, M.E., Cammas, S., Appel, M., Ponchel, G. 2004. Investigation of the degradation 549

mechanisms of poly(malic acid) esters in vitro and their related cytotoxicities on J774 macrophages., 550

Biomacromolecules 5, 137-143. 551

552

Mazzaferro, V., Chun, Y.S., Poon, R.T., Schwartz, M.E., Yao, F.Y., Marsh, J.W., Bhoori, S., Lee, S.G. 553

2008. Liver Transplantation for Hepatocellular Carcinoma., Ann. Surg. Oncol. 15(4), 1001-1007. 554

555

Misra, R., Acharya, S., Sahoo, S.K. 2010. Cancer nanotechnology: application of nanotechnology in 556

cancer therapy., Drug Discovery Today 15(19/20), 842-850. 557

558

Osanai, S., Nakamura, K. 2000. Effect of complexation between liposome and poly(malic acid) on 559

aggregation and leakage behaviour., Biomaterials 21, 867-876. 560

561

Parkin, D.M., Bray, F., Ferlay, J., Pisani, P. 2005. Global Cancer Statistics, 2002. CA. Cancer J. Clin. 562

55(2), 74-108. 563

564

Patil, Y.B., Toti, U.S., Khdair, A., Ma, L., Panyam, J. 2009. Single-step surface functionalization of 565

polymeric nanoparticles for targeted drug delivery., Biomaterials 30, 859-866. 566

(24)

23 Psimadas, D., Georgoulias, P., Valotassiou, V., Loudos, G. 2012. Molecular Nanomedicine Towards 568

Cancer: 111In-labeled Nanoparticles., J. Pharm. Sci. 101(7), 2271-2280. 569

570

Rahbari, R., Sheahan, T., Modes, V., Collier, P., Macfarlane, C., Badge, R.M. 2009. A novel L1 571

retrotransposon marker for HeLa cell line identification., BioTechniques 46(4), 277–284. 572

573

Reddy, L.H., Couvreur, P. 2011. Nanotechnology for therapy and imaging of liver diseases., J. 574

Hepatology 55(6), 1461-1466. 575

576

Romberg, B., Hennink, W.E., Strom, G. 2008. Sheddable coatings for long-circulating nanoparticles., 577

Pharmaceutical Research 25(1), 55-71. 578

579

Thioune, O., Fessi, H., Devissaguet, J.P., Puisieux, F. 1997. Preparation of pseudolatex by 580

nanoprecipitation: influence of the solvent nature on intrinsic viscosity and interaction constant., Int. J. 581

Pharm. 146, 233-238. 582

583

Vert, M., Lenz, R.W. 1979. Preparation and properties of polyβ-malic acid: a functional polyester of 584

potential biomedical importance., Polym. Prepr. (Am. Chem. Soc., Div. Polym. Chem.) 20(1), 608-585

611. 586

587

Wang, A.Z., Langer, R., Farokhzad, O.C. 2012. Nanoparticle Delivery of Cancer Drugs., Annu. Rev. 588

Med. 63, 185-198. 589

590

Wu, J., Nantz, M.H., Zern, M.A. 2002. Targeting Hepatocytes for Drug and Gene Delivery: Emerging 591

Novel Approaches and Applications., Front. Biosci. 7, 717-725. 592

(25)

24 Yan, Y., Such, G.K., Johnston, A.P.R., Best, J.P., Caruso, F. 2012. Engineering Particles for 594

Therapeutic Delivery: Prospects and Challenges., ACS Nano. 6(5), 3663-3669. 595

596

Yang, W., Cheng, Y., Xu, T., Wang, X., Wen, L.P. 2009. Targeting cancer cells with biotin-dendrimer 597

conjugates., Eur. J. Med. Chem. 44, 862-868. 598

(26)

25 Table 1. Characteristics of Dox-loaded NPs.

600 NPs Diameter (nm)a Polydispersity index (Ip)a Zeta potential (mV)a Encapsulation efficiency (%)b PEG42-b-PMLABe 66 ± 7 0.22 - 8 ± 2 36 ± 3 Biot-PEG62-b-PMLABe 74 ± 4 0.22 - 6 ± 1 32 ± 2

a. Measured by DLS (Delsa™ Nano Beckman Coulter); b. Measured by UV at 485 nm.

601 602

(27)

26 Table 2. Characteristics of fluorescent NPs measured by DLS (Delsa™ Nano Beckman Coulter). 603

NPs Diameter (nm) Polydispersity index

PEG42-b-PMLABe-FA + PEG42-b-PMLABe (50wt%) 64 ± 14 0.16

Biot-PEG62-b-PMLABe-FA + Biot-PEG62-b-PMLABe (50wt%) 70 ± 13 0.22

604 605 606 607 608 609 610 611 612 613 614 615 616 617

(28)

27 Table 3. Characteristics of NPs modified or not by the strep-Biot-RGD construct measured by DLS 618

(Delsa™ Nano Beckman Coulter). 619 Conditions NPs Biot-PEG62-b-PMLABe NPs Biot-PEG62-b-PMLABe + strep-Biot-RGD Diameter (nm) Ip Diameter (nm) Ip Initial (2.5 g/L) 53 ± 12 0.17 -- --

Solution in PBS (0.107 g/L) 79 ± 15 0.37 208 ± 30 (a) 0.35(a)

Solution in culture medium without serum (0.107 g/L) 78 ± 15 0.29 191 ± 21 0.28

Solution in culture medium with serum (0.107 g/L) 106 ± 30 0.15 185 ± 30 (b) 0.23(b)

(a). Presence of a second peak centered at 600 ± 100 nm; (b). Presence of a second peak centered at 620

970 ± 220 nm 621

(29)

28 Figure captions:

623

Figure 1. Uses of PMLA derivatives as building blocks for the design of versatile NPs. 624

625

Figure 2. Release profile of Dox in water (A) and in culture medium (B) at 37°C. 626

627

Figure 3. Schematic representation of the molecular scaffold following grafting of biotinylated peptide 628

on the biotinylated NPs via streptavidin as a bridging factor. 629

630

Figure 4. Flow cytometry (right) and in situ fluorescence microscopy (left) analysis of HepaRG cells 631

incubated with Biot-PEG62-b-PMLABe NPs, Biot-PEG62-b-PMLABe-FA NPs and

RGD-Biot-Strept-632

Biot-PEG62-b-PMLABe-FA NPs. For Facs analysis, size (FSC-H) and granularity (SSC-H) were

633

visualized to select the R1 gate corresponding to living cells. Detection on fluorescent cells in the R1 634

gate was performed using the FL1-H channel: negative cells incubated with Biot-PEG62-b-PMLABe

635

NPs were set in the M1 window and positive cells were detected in the M2 window. For cells 636

incubated with Biot-PEG62-b-PMLABe-FA and RGD-Biot-Strept-Biot-PEG62-b-PMLABe-FA NPs (1)

637

50 and (1) 90% of cells were fluorescent, respectively; white bar : 50 µm. 638

639

Figure 5. Time course of the uptake of NPs by HepaRG cells. Red curve: RGD-Biot-Strept-Biot-640

PEG62-b-PMLABe-FA NPs; Blue curve: Biot-PEG62-b-PMLABe-FA NPs; white bar : 50 µm.

641 642

Figure 6. Captation of fluorescent nanoparticles (24 hour incubation) by HepaRG cells in absence or in 643

presence of free RGD peptide; Flow cytometry (Left) and fluorescence microscopy (Rigth) analyses 644

with Biot-PEG62-b-PMLABe-FA NPs, RGD-Biot-Strept-Biot-PEG62-b-PMLABe-FA NPs and

RGD-645

Biot-Strept-Biot-PEG62-b-PMLABe-FA NPs + 34 µM of free RGD peptide. Bottom chart: Mean of

646

fluorescence under the various tested conditions. 647

(30)

29 Figure 7. Flow cytometry measurements of fluorescence in HepaRG, HT29 and HeLa cells incubated 648

with Biot-PEG62-b-PMLABe-FA or RGD-Biot-Strept-Biot-PEG62-b-PMLABe-FA formed NPs for 24

649

hours. Left chart: Number of positive cells; right chart: Mean of fluorescence. 650

(31)

30 Figure 1. 652 653 654 PEG42-b-PMLABe Biot-PEG62-b-PMLABe PEG42-b-PMLABe-FA Biot-PEG62-b-PMLABe-FA Nanoprecipitation Dox ( )

PEG42-b-PMLABe [Dox] NPs

Biot-PEG62-b-PMLABe[Dox] NPs

Dialysis against H2O

or culture medium Dox release profile

Nanoprecipitation PEG42-b-PMLABe-FA NPs Biot-PEG62-b-PMLABe-FA NPs RGD-Biot-Strept-Biot-PEG62-b-PMLABe NPs RGD-Biot-Strept NPs’ uptake in vitro Biotin Streptavidin (Strept) RGD peptide

(32)

31 Figure 2. 655 0% 10% 20% 30% 40% 50% 60% 70% 80% 90% 100% 0 10 20 30 40 50 60 70 80 R e le a se r a te ( % )

Incubation time (hours)

B PEG-b-PMLABe[Dox] NPs Biot-PEG-b-PMLABe[Dox] NPs 0% 10% 20% 30% 40% 50% 60% 70% 80% 90% 100% 0 10 20 30 40 50 60 70 80 R e le a se r a te ( % )

Incubation time (hours)

A

PEG-b-PMLABe[Dox] NPs Biot-PEG-b-PMLABe[Dox] NPs

656 657

(33)

32 Figure 3. 658 659 660 661 662 663 664 665 123 4567 879AB7CD5E5F 2AB75EA AAB769 A 9

(34)

33 Figure 4.

666

667 668

HepaRG control neg.001

0 200 400 600 800 1000 FSC-H R1 HepaRG PMLAbiotFA.002 0 200 400 600 800 1000 FSC-H R1 HepaRG PMLAbiotFARGD c3.003 0 200 400 600 800 1000 FSC-H R1 Biot-PEG62-b-PMLABe NPs E v e n ts E v e n ts E v e n ts S S C -H S S C -H S S C -H Biot-PEG62-b-PMLABe-FA NPs RGD-Biot-PEG62-b-PMLABe-FA NPs HepaRG control neg.001

100 101 102 103 104 FL1-H M1 M2 HepaRG PMLAbiotFA.002 100 101 102 103 104 FL1-H M1 M2 100 101 102 103 104 FL1-H M1 M2

(35)

34 Figure 5. 669 670 671 672 673 674 675 676 677 0 50 100 150 200 250 300 350 400 0 6 12 18 24 Biot-PEG62-b-PMLABe-FA NPs RGD-Biot-PEG62-b-PMLABe-FA NPs Time (hours) F lu o re sc e n c e i n te n si ty ( A U )

(36)

35 Figure 6. 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 100 101 102 103 104 FL1-H M1 M2

HepRG PMLAFAbiot RGDcond3c.009

100 101 102 103 104 FL1-H M1 M2 100 101 102 103 104 FL1-H M1 M2 Biot-PEG62-b-PMLABe-FA NPs RGD-Biot-PEG62-b-PMLABe-FA NPs

RGD-Biot-PEG62-b-PMLABe-FA NPs + free RGD

E v e n ts E v e n ts E v e n ts 1 211 311 411 511 611 711 811 2 3 4 5 M e a n o f fl u o re s c e n c e ( U .A .)

(37)

36 Figure 7. 704 705 706 100 101 102 103 104 FL1-H M1 M2 100 101 102 103 104 FL1-H M1 M2

HeLa PMLA biotFA RGDc3.009

100 101 102 103 104 FL1-H M1 M2 1 31 51 71 91 211 2 3 4 5 6 7 8 9 A 1 211 311 411 511 611 2 3 4 5 6 7 8 9 A Me a n o f fl u o re s c e n c e ( U .A .) P o s it iv e c e ll s ( % )

HepaRG HT29 HeLa HepaRG HT29 HeLa

HeLa

HepaRG neg cond 1a.001

100 101 102 103 104 FL1-H

M1

M2

HepaRG PMLAFAbiot cond 2a.004

100 101 102 103 104 FL1-H

M1

M2

HepRG PMLAFAbiot RGDcond3c.009

100 101 102 103 104 FL1-H M1 M2 HepaRG Without NPs 100 101 102 103 104 FL1-H M1 M2 HT29 PMLA FA RGD c3.015 100 101 102 103 104 FL1-H M1 M2 HT29 PMLA FAc2.014 100 101 102 103 104 FL1-H M1 M2 HT29

(38)

37 Scheme captions

707 708

Scheme 1. Synthetic route to FA grafted PEG42-b-PMLABe and Biot-PEG62-b-PMLABe. 709

(39)

38 Scheme 1. 711 712 R b CH CH2 CO2CH2C6H5 C O O H n R: PEG42-; PEG42-b-PMLABe

R: Biot-PEG62-; Biot-PEG62-b-PMLABe

DCC/NHS CH2Cl2anhydrous RT, 24 h R b CH CH2 CO2CH2C6H5 C O O NSH n FA CH2Cl2anhydrous RT, 24 h R b CH CH2 CO2CH2C6H5 C O O FA n R: PEG42-; PEG42-b-PMLABe-FA

R: Biot-PEG62-; Biot-PEG62-b-PMLABe-FA O O OH HO H2N O Fluorescein amine, FA

Références

Documents relatifs

actual thrust contact between the two rock types is located in a zone of intensely deformed veined calcite, referred to in the literature as Lochsitekalk (limestone) or

As numerical simulations make possible to deduce the characteristics of the phenomena in real size, reduced scale physical emulation based on HIL experiments minimizes the

lizing algorithm building local routing tables under the fair daemon (the tables ensure the routing from any node v to its t closest nodes) in O(D) rounds in the connected component,

/ La version de cette publication peut être l’une des suivantes : la version prépublication de l’auteur, la version acceptée du manuscrit ou la version de l’éditeur.. Access

A Reactive Walking Pattern Generator Based on Nonlinear Model Predictive Control.. Maximilien Naveau, Manuel Kudruss, Olivier Stasse, Christian Kirches, Katja Mombaur,

Access and use of this website and the material on it are subject to the Terms and Conditions set forth at Some unusual insects in houses..

We propose in this section a review of evidence- based studies conducted in West Africa and abroad that have emphasized a wide range of digital interventions in four

Keywords: epilepsy surgery; drug-resistant epilepsy; neurosurgery; shear-wave elastography; real- time guidance; epileptogenic zone; brain surgery; focal epilepsy; ultrasound;